Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Front Immunol ; 14: 1254762, 2023.
Article in English | MEDLINE | ID: mdl-37908354

ABSTRACT

Schistosomiasis-associated Pulmonary Arterial Hypertension (Sch-PAH) is a life-threatening complication of chronic S. mansoni infection that can lead to heart failure and death. During PAH, the expansion of apoptosis-resistant endothelial cells (ECs) has been extensively reported; however, therapeutic approaches to prevent the progression or reversal of this pathological phenotype remain clinically challenging. Previously, we showed that depletion of the anti-apoptotic protein Caveolin-1 (Cav-1) by shedding extracellular vesicles contributes to shifting endoprotective bone morphogenetic protein receptor 2 (BMPR2) towards transforming growth factor beta (TGF-ß)-mediated survival of an abnormal EC phenotype. However, the mechanism underlying the reduced endoprotection in PAH remains unclear. Interestingly, recent findings indicate that, similar to the gut, healthy human lungs are populated by diverse microbiota, and their composition depends significantly on intrinsic and extrinsic host factors, including infection. Despite the current knowledge that the disruption of the gut microbiome contributes to the development of PAH, the role of the lung microbiome remains unclear. Thus, using a preclinical animal model of Sch-PAH, we tested whether S. mansoni infection alters the gut-lung microbiome composition and causes EC injury, initiating the expansion of an abnormal EC phenotype observed in PAH. Indeed, in vivo stimulation with S. mansoni eggs significantly altered the gut-lung microbiome profile, in addition to promoting injury to the lung vasculature, characterized by increased apoptotic markers and loss of endoprotective expression of lung Cav-1 and BMPR2. Moreover, S. mansoni egg stimulus induced severe pulmonary vascular remodeling, leading to elevated right ventricular systolic pressure and hypertrophy, characteristic of PAH. In vitro, exposure to the immunodominant S. mansoni egg antigen p40 activated TLR4/CD14-mediated transient phosphorylation of Cav-1 at Tyr14 in human lung microvascular EC (HMVEC-L), culminating in a mild reduction of Cav-1 expression, but failed to promote death and shedding of extracellular vesicles observed in vivo. Altogether, these data suggest that disruption of the host-associated gut-lung microbiota may be essential for the emergence and expansion of the abnormal lung endothelial phenotype observed in PAH, in addition to S. mansoni eggs and antigens.


Subject(s)
Gastrointestinal Microbiome , Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Schistosomiasis , Animals , Mice , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Caveolin 1/genetics , Endothelial Cells/metabolism , Hypertension, Pulmonary/etiology , Lung/pathology , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Schistosomiasis/metabolism
2.
Pulm Circ ; 12(4): e12163, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36484056

ABSTRACT

Dysfunctional bone morphogenetic protein receptor 2 (BMPR2) and endothelial nitric oxide synthase (eNOS) have been largely implicated in the pathogenesis of pulmonary arterial hypertension (PAH); a life-threatening cardiopulmonary disease. Although the incident of PAH is about three times higher in females, males with PAH usually have a worse prognosis, which seems to be dependent on estrogen-associated cardiac and vascular protection. Here, we evaluated whether hypoxia-induced pulmonary hypertension (PH) in humanized BMPR2+/R899X loss-of-function mutant mice contributes to sex-associated differences observed in PAH by altering eNOS expression and inducing expansion of hyperactivated TGF-ß-producing pulmonary myofibroblasts. To test this hypothesis, male and female wild-type (WT) and BMPR2+/R899X mutant mice were kept under hypoxic or normoxic conditions for 4 weeks, and then right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH) were measured. Chronic hypoxia exposure elevated RVSP, inducing RVH in both groups, with a greater effect in BMPR2+/R899X female mice. Lung histology revealed no differences in vessel thickness/area between sexes, suggesting RVSP differences in this model are unlikely to be in response to sex-dependent vascular narrowing. On the other hand, hypoxia exposure increased vascular collagen deposition, the number of TGF-ß-associated α-SMA-positive microvessels, and eNOS expression, whereas it also reduced caveolin-1 expression in the lungs of BMPR2+/R899X females compared to males. Taken together, this brief report reveals elevated myofibroblast-derived TGF-ß and eNOS-derived oxidants contribute to pulmonary microvascular muscularization and sex-linked differences in incidence, severity, and outcome of PAH.

3.
Arterioscler Thromb Vasc Biol ; 42(9): 1113-1120, 2022 09.
Article in English | MEDLINE | ID: mdl-35861068

ABSTRACT

Over the last 2 years, the COVID-19 pandemic has stimulated the scientific community by starting a race to develop new vaccines and therapeutic approaches to fight this life-threatening illness. At the same time, the pandemic also evoked an urge for innovative communication strategies to maintain scientific networking and data sharing among investigators. Communication through audiovisual platforms has quickly become a unique tool to sustain scientific interaction, whereas social media has turned into an unmistakable pivotal environment for sharing scientific data and combating misinformation around SARS-CoV-2 infection, prevention, and therapy. Amid this challenging scenario, the scientific community organically established new roles, such as a social media ambassador, a conference-associated role to virtually promote breakthrough science while reconnecting investigators and forging new scientific networks via social media. Moreover, in response to the COVID-19 pandemic, it also became clear the critical need for the scientific community to support efforts to empower flexibility, creativity, and the inclusion of new forms of communication to advance science. Thus, the goal of this brief article is to provide a structured follow-up on the importance for researchers to occupy the internet to promote scientific findings and events, to combat science mistrust by stimulating communication among nonscientists to scientists, and to provide essential strategies for young and senior investigators on how to virtually expand their professional networks within and across research and clinical areas of the cardiovascular field.


Subject(s)
COVID-19 , Social Media , Communication , Humans , Pandemics , SARS-CoV-2
4.
Biomed Pharmacother ; 149: 112784, 2022 May.
Article in English | MEDLINE | ID: mdl-35299122

ABSTRACT

Chalcones (1,3-diphenyl-2-propen-1-ones) either natural or synthetic have a plethora of biological properties including antileishmanial activities, but their development as drugs is hampered by their largely unknown mechanisms of action. We demonstrate herein that our previously described benzochalcone fluorogenic probe (HAB) could be imaged by fluorescence microscopy in live Leishmania amazonensis promastigotes where it targeted the parasite acidocalcisomes, lysosomes and the mitochondrion. As in the live zebrafish model, HAB formed yellow-emitting fluorescent complexes when associated with biological targets in Leishmania. Further, we used HAB as a reversible probe to study the binding of a portfolio of diverse chalcones and analogues in live promastigotes, using a combination of competitive flow cytometry analysis and cell microscopy. This pharmacological evaluation suggested that the binding of HAB in promastigotes was representative of chalcone pharmacology in Leishmania, with certain exogenous chalcones exhibiting competitive inhibition (ca. 20-30%) towards HAB whereas non-chalconic inhibitors showed weak capacity (ca. 3-5%) to block the probe intracellular binding. However, this methodology was restricted by the strong toxicity of several competing chalcones at high concentration, in conjunction with the limited sensitivity of the HAB fluorophore. This advocates for further optimization of this undirect target detection strategy using pharmacophore-derived reversible fluorescent probes.


Subject(s)
Antiprotozoal Agents , Chalcone , Chalcones , Leishmania , Animals , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Binding Sites , Chalcone/pharmacology , Chalcones/chemistry , Chalcones/pharmacology , Fluorescent Dyes , Zebrafish
5.
J Cell Sci ; 133(5)2020 03 06.
Article in English | MEDLINE | ID: mdl-32005701

ABSTRACT

Macrophages are tissue-resident immune cells that are crucial for the initiation and maintenance of immune responses. Purinergic signaling modulates macrophage activity and impacts cellular plasticity. The ATP-activated purinergic receptor P2X7 (also known as P2RX7) has pro-inflammatory properties, which contribute to macrophage activation. P2X7 receptor signaling is, in turn, modulated by ectonucleotidases, such as CD39 (also known as ENTPD1), expressed in caveolae and lipid rafts. Here, we examined P2X7 receptor activity and determined impacts on ectonucleotidase localization and function in macrophages primed with lipopolysaccharide (LPS). First, we verified that ATP boosts CD39 activity and caveolin-1 protein expression in LPS-primed macrophages. Drugs that disrupt cholesterol-enriched domains - such as nystatin and methyl-ß-cyclodextrin - decreased CD39 enzymatic activity in all circumstances. We noted that CD39 colocalized with lipid raft markers (flotillin-2 and caveolin-1) in macrophages that had been primed with LPS followed by treatment with ATP. P2X7 receptor inhibition blocked these ATP-mediated increases in caveolin-1 expression and inhibited the colocalization with CD39. Further, we found that STAT3 activation is significantly attenuated caveolin-1-deficient macrophages treated with LPS or LPS+BzATP. Taken together, our data suggest that P2X7 receptor triggers the initiation of lipid raft-dependent mechanisms that upregulates CD39 activity and could contribute to limit macrophage responses restoring homeostasis.


Subject(s)
Caveolin 1 , Receptors, Purinergic P2X7 , Adenosine Triphosphate , Caveolin 1/genetics , Lipopolysaccharides , Macrophages , Membrane Microdomains , Receptors, Purinergic P2X7/genetics
6.
Anesthesiology ; 131(6): 1301-1315, 2019 12.
Article in English | MEDLINE | ID: mdl-31658116

ABSTRACT

BACKGROUND: Sevoflurane with its antiinflammatory properties has shown to decrease mortality in animal models of sepsis. However, the underlying mechanism of its beneficial effect in this inflammatory scenario remains poorly understood. Macrophages play an important role in the early stage of sepsis as they are tasked with eliminating invading microbes and also attracting other immune cells by the release of proinflammatory cytokines such as interleukin-1ß, interleukin-6, and tumor necrosis factor-α. Thus, the authors hypothesized that sevoflurane mitigates the proinflammatory response of macrophages, while maintaining their bactericidal properties. METHODS: Murine bone marrow-derived macrophages were stimulated in vitro with lipopolysaccharide in the presence and absence of 2% sevoflurane. Expression of cytokines and inducible NO synthase as well as uptake of fluorescently labeled Escherichia coli (E. coli) were measured. The in vivo endotoxemia model consisted of an intraperitoneal lipopolysaccharide injection after anesthesia with either ketamine and xylazine or 4% sevoflurane. Male mice (n = 6 per group) were observed for a total of 20 h. During the last 30 min fluorescently labeled E. coli were intraperitoneally injected. Peritoneal cells were extracted by peritoneal lavage and inducible NO synthase expression as well as E. coli uptake by peritoneal macrophages was determined using flow cytometry. RESULTS: In vitro, sevoflurane enhanced lipopolysaccharide-induced inducible NO synthase expression after 8 h by 466% and increased macrophage uptake of fluorescently labeled E. coli by 70% compared with vehicle-treated controls. Inhibiting inducible NO synthase expression pharmacologically abolished this increase in bacteria uptake. In vivo, inducible NO synthase expression was increased by 669% and phagocytosis of E. coli by 49% compared with the control group. CONCLUSIONS: Sevoflurane enhances phagocytosis of bacteria by lipopolysaccharide-challenged macrophages in vitro and in vivo via an inducible NO synthase-dependent mechanism. Thus, sevoflurane potentiates bactericidal and antiinflammatory host-defense mechanisms in endotoxemia.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation, Enzymologic , Macrophages/enzymology , Nitric Oxide Synthase Type II/biosynthesis , Phagocytosis/physiology , Sevoflurane/pharmacology , Animals , Blood Bactericidal Activity/drug effects , Blood Bactericidal Activity/physiology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Phagocytosis/drug effects , RAW 264.7 Cells
7.
Chem Sci ; 10(12): 3654-3670, 2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30996961

ABSTRACT

Neutrophil granules (NGs) are key components of the innate immune response and mark the development of neutrophilic granulocytes in mammals. However, there has been no specific fluorescent vital stain up to now to monitor their dynamics within a whole live organism. We rationally designed a benzochalcone fluorescent probe (HAB) featuring high tissue permeability and optimal photophysics such as elevated quantum yield, pronounced solvatochromism and target-induced fluorogenesis. Phenotypic screening identified HAB as the first cell- and organelle-specific small-molecule fluorescent tracer of NGs in live zebrafish larvae, with no labeling of other cell types or organelles. HAB staining was independent of the state of neutrophil activation, labeling NGs of both resting and phagocytically active neutrophils with equal specificity. By high-resolution live imaging, we documented the dynamics of HAB-stained NGs during phagocytosis. Upon zymosan injection, labeled NGs were rapidly recruited to the forming phagosomes. Despite being a reversible ligand, HAB could not be displaced by high concentrations of pharmacologically relevant competing chalcones, indicating that this specific labeling was the result of the HAB's precise physicochemical signature rather than a general feature of chalcones. However, one of the competitors was discovered as a promising interstitial fluorescent tracer illuminating zebrafish histology, similarly to BODIPY-ceramide. As a yellow-emitting histopermeable vital stain, HAB functionally and spectrally complements most genetically incorporated fluorescent tags commonly used in live zebrafish biology, holding promise for the study of neutrophil-dependent responses relevant to human physiopathology such as developmental defects, inflammation and infection. Furthermore, HAB intensely labeled isolated live human neutrophils at the level of granulated subcellular structures consistent with human NGs, suggesting that the labeling of NGs by HAB is not restricted to the zebrafish model but also relevant to mammalian systems.

8.
Arterioscler Thromb Vasc Biol ; 39(6): 1191-1202, 2019 06.
Article in English | MEDLINE | ID: mdl-30943774

ABSTRACT

Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-ß (transforming growth factor-ß) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-ß production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-ß, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-ß-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-ß signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.


Subject(s)
Caveolin 1/deficiency , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Pulmonary Arterial Hypertension/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Adolescent , Adult , Aged , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Caveolin 1/genetics , Cell Proliferation , Disease Models, Animal , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Female , Humans , Hypoxia/complications , Indoles , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/pathology , Pyrroles , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , Young Adult
9.
Curr Top Membr ; 82: 257-279, 2018.
Article in English | MEDLINE | ID: mdl-30360781

ABSTRACT

Pulmonary vascular diseases are associated with several factors including infection, cigarette smoking, abuse of dietary suppressants and drugs, prolonged exposure to high altitude, and other causes which in part induce significant oxidative stress resulting in endothelial cell injury, apoptosis, hyperproliferation, and vaso-occlusive disease. Maintenance of normal endothelial cell function is a critical role of endothelial nitric oxide synthase (eNOS) activity and physiologic nitric oxide (NO) signaling in the vascular wall. eNOS expression and activity is regulated by the membrane-associated scaffolding protein caveolin-1 (Cav-1), the main protein constituent of caveolae. This chapter summarizes the literature and highlights unanswered questions related to how inflammation-associated oxidative stress affects Cav-1 expression and regulatory functions, and how dysregulated eNOS enzymatic activity promotes endothelial dysfunction. Focus is given to how the conversion of eNOS from a NO-producing enzyme to a transient oxidant-generating system is associated twith Cav-1 depletion, endothelial cell injury, and pulmonary vascular diseases. Importantly, the vascular defects observed in absence of Cav-1 that give rise to injured or hyperproliferative endothelial cells and promote remodeled vasculature can be rescued by "re-coupling," inhibiting, or genetically deleting eNOS, supporting the notion that strict control of Cav-1 expression and eNOS activity and signaling is critical for maintaining pulmonary vascular homeostasis.


Subject(s)
Caveolin 1/metabolism , Nitric Oxide/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Lung/cytology , Lung/metabolism , Mechanotransduction, Cellular , Nitric Oxide Synthase Type III/metabolism , Signal Transduction
10.
Free Radic Biol Med ; 112: 587-596, 2017 11.
Article in English | MEDLINE | ID: mdl-28899725

ABSTRACT

[Ru(terpy)(bdq)NO]3+ (TERPY) is a nitric oxide (NO) donor that promotes relaxation of the mesenteric artery and aorta in rats. We sought to investigate whether it acts as both an NO donor and endothelial NO synthase (eNOS) activator, as shown previously for nitroglycerin. Human umbilical vein endothelial cells (HUVECs) and human embryonic kidney 293 cells transfected with empty vector (HEK) or eNOS cDNA (HEK-eNOS) were treated with TERPY (1µM) for different lengths of time. eNOS expression, dimerization, and Ser1177 phosphorylation, caveolin-1 (Cav-1) oligomerization, Cav-1 Tyr14 phosphorylation were evaluated by Western blotting. Studies also assessed the production of reactive oxygen/nitrogen species (ROS/RNS) in HUVECs and HEK-eNOS cells. In HEK cells devoid of eNOS, TERPY released NO without additional stimulus indicating that is an NO donor. Moreover, in HEK-eNOS cells, TERPY-induced NO production that was blocked by L-NAME. In addition, TERPY increased ROS and ONOO- production which were blocked by more than 80% by BH4 (essential eNOS co-factor) and eNOS siRNA. These results suggest that TERPY-induced ROS and ONOO- production were originated from eNOS. HUVECs stimulated with TERPY showed increased eNOS Ser1177 and Cav-1 Tyr14 phosphorylation, and decreased eNOS dimerization, Cav-1 oligomerization, and Cav-1/eNOS interaction after 20min. It suggests that TERPY induces eNOS hyperactivation and uncoupling by disrupting Cav-1/eNOS interaction and depleting BH4. Endothelium-dependent vasodilation in response to NO donor TERPY is associated with eNOS activation and uncoupling, and thereby appears to be mediated, at least in part, via eNOS-dependent ROS/RNS production.


Subject(s)
Caveolin 1/genetics , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/biosynthesis , Organometallic Compounds/pharmacology , Biopterins/analogs & derivatives , Biopterins/pharmacology , Caveolin 1/metabolism , Fluoresceins , Fluorescent Dyes , Gene Expression , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/agonists , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects , Plasmids/chemistry , Plasmids/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Spectrometry, Fluorescence , Transfection
11.
J Hepatol ; 67(4): 716-726, 2017 10.
Article in English | MEDLINE | ID: mdl-28554875

ABSTRACT

BACKGROUND & AIMS: The severity of sepsis can be linked to excessive inflammatory responses resulting in hepatic injury. P2X7 receptor activation by extracellular ATP (eATP) exacerbates inflammation by augmenting cytokine production; while CD39 (ENTPD1) scavenges eATP to generate adenosine, thereby limiting P2X7 activation and resulting in A2A receptor stimulation. We aim to determine how the functional interaction of P2X7 receptor and CD39 control the macrophage response, and consequently impact on sepsis and liver injury. METHODS: Sepsis was induced by cecal ligation and puncture in C57BL/6 wild-type (WT) and CD39-/- mice. Several in vitro assays were performed using peritoneal or bone marrow derived macrophages to determine CD39 ectonucleotidase activity and its role in sepsis-induced liver injury. RESULTS: CD39 expression in macrophages limits ATP-P2X7 receptor pro-inflammatory signaling. P2X7 receptor paradoxically boosts CD39 activity. Inhibition and/or deletion of P2X7 receptor in LPS-primed macrophages attenuates cytokine production and inflammatory signaling as well as preventing ATP-induced increases in CD39 activity. Septic CD39-/- mice exhibit higher levels of inflammatory cytokines and show more pronounced liver injury than WT mice. Pharmacological P2X7 blockade largely prevents tissue damage, cell apoptosis, cytokine production, and the activation of inflammatory signaling pathways in the liver from septic WT, while only attenuating these outcomes in CD39-/- mice. Furthermore, the combination of P2X7 blockade with adenosine A2A receptor stimulation completely inhibits cytokine production, the activation of inflammatory signaling pathways, and protects septic CD39-/- mice against liver injury. CONCLUSIONS: CD39 attenuates sepsis-associated liver injury by scavenging eATP and ultimately generating adenosine. We propose boosting of CD39 would suppress P2X7 responses and trigger adenosinergic signaling to limit systemic inflammation and restore liver homeostasis during the acute phase of sepsis. Lay summary: CD39 expression in macrophages limits P2X7-mediated pro-inflammatory responses, scavenging extracellular ATP and ultimately generating adenosine. CD39 genetic deletion exacerbates sepsis-induced experimental liver injury. Combinations of a P2X7 antagonist and adenosine A2A receptor agonist are hepatoprotective during the acute phase of abdominal sepsis.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Liver/immunology , Liver/injuries , Receptors, Purinergic P2X7/metabolism , Sepsis/immunology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Apyrase/deficiency , Apyrase/genetics , Cytokines/biosynthesis , Disease Models, Animal , Interleukin-1beta/biosynthesis , Liver/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/deficiency , Receptors, Purinergic P2X7/genetics , STAT3 Transcription Factor/metabolism , Sepsis/therapy , Signal Transduction/drug effects , Signal Transduction/immunology
12.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L760-L771, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28188225

ABSTRACT

Endothelial cell (EC) activation and vascular injury are hallmark features of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Caveolin-1 (Cav-1) is highly expressed in pulmonary microvascular ECs and plays a key role in maintaining vascular homeostasis. The aim of this study was to determine if the lung inflammatory response to Escherichia coli lipopolysaccharide (LPS) promotes priming of ECs via Cav-1 depletion and if this contributes to the onset of pulmonary vascular remodeling. To test the hypothesis that depletion of Cav-1 primes ECs to respond to profibrotic signals, C57BL6 wild-type (WT) mice (Tie2.Cre-;Cav1fl/fl ) were exposed to nebulized LPS (10 mg; 1 h daily for 4 days) and compared with EC-specific Cav1-/- (Tie2.Cre+;Cav1fl/fl ). After 96 h of LPS exposure, total lung Cav-1 and bone morphogenetic protein receptor type II (BMPRII) expression were reduced in WT mice. Moreover, plasma albumin leakage, infiltration of immune cells, and levels of IL-6/IL-6R and transforming growth factor-ß (TGF-ß) were elevated in both LPS-treated WT and EC-Cav1-/- mice. Finally, EC-Cav1-/- mice exhibited a modest increase in microvascular thickness basally and even more so on exposure to LPS (96 h). EC-Cav1-/- mice and LPS-treated WT mice exhibited reduced BMPRII expression and endothelial nitric oxide synthase uncoupling, which along with increased TGF-ß promoted TGFßRI-dependent SMAD-2/3 phosphorylation. Finally, human lung sections from patients with ARDS displayed reduced EC Cav-1 expression, elevated TGF-ß levels, and severe pulmonary vascular remodeling. Thus EC Cav-1 depletion, oxidative stress-mediated reduction in BMPRII expression, and enhanced TGF-ß-driven SMAD-2/3 signaling promote pulmonary vascular remodeling in inflamed lungs.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Caveolin 1/metabolism , Endothelial Cells/pathology , Inflammation/pathology , Lung/blood supply , Lung/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Actins/metabolism , Acute Lung Injury/complications , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Adult , Aged , Animals , Bronchoalveolar Lavage Fluid , Cell Shape/drug effects , Cytokines/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-6/pharmacology , Lipopolysaccharides , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Nitric Oxide Synthase Type III/metabolism , Proteolysis/drug effects , Pulmonary Artery/pathology , Receptors, Transforming Growth Factor beta/metabolism , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Time Factors , Vascular Remodeling/drug effects
13.
Mediators Inflamm ; 2014: 134974, 2014.
Article in English | MEDLINE | ID: mdl-25276050

ABSTRACT

Schistosomiasis is a chronic inflammatory disease whose macrophages are involved in immunopathology modulation. Although P2X7 receptor signaling plays an important role in inflammatory responses mediated by macrophages, no reports have examined the role of P2X7 receptors in macrophage function during schistosomiasis. Thus, we evaluated P2X7 receptor function in peritoneal macrophages during schistosomiasis using an ATP-induced permeabilization assay and measurements of the intracellular Ca(2+) concentration. ATP treatment induced significantly less permeabilization in macrophages from S. mansoni-infected mice than in control cells from uninfected animals. Furthermore, P2X7-mediated increases in intracellular Ca(2+) levels were also reduced in macrophages from infected mice. TGF-ß1 levels were increased in the peritoneal cavity of infected animals, and pretreatment of control macrophages with TGF-ß1 reduced ATP-induced permeabilization, mimicking the effect of S. mansoni infection. Western blot and qRT-PCR data showed no difference in P2X7 protein and mRNA between uninfected, infected, and TGF-ß1-treated groups. However, immunofluorescence analysis revealed reduced cell surface localization of P2X7 receptors in macrophages from infected and TGF-ß1-treated mice compared to controls. Therefore, our data suggest that schistosomiasis reduces peritoneal macrophage P2X7 receptor signaling. This effect is likely due to the fact that infected mice have increased levels of TGF-ß1, which reduces P2X7 receptor cell surface expression.


Subject(s)
Macrophages, Peritoneal/metabolism , Receptors, Purinergic P2X7/metabolism , Schistosomiasis/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Male , Mice , Mice, Knockout , Microscopy, Confocal , Receptors, Purinergic P2X7/genetics , Reverse Transcriptase Polymerase Chain Reaction
14.
Toxicon ; 67: 55-62, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23474269

ABSTRACT

In this work we evaluated the ability of suramin, a polysulfonated naphthylurea derivative, to antagonize the cytotoxic and enzymatic effects of the crude venom of Apis mellifera. Suramin was efficient to decrease the lethality in a dose-dependent way. The hemoconcentration caused by lethal dose injection of bee venom was abolished by suramin (30 µg/g). The edematogenic activity of the venom (0.3 µg/g) was antagonized by suramin (10 µg/g) in all treatment protocols. The changes in the vascular permeability caused by A. mellifera (1 µg/g) venom were inhibited by suramin (30 µg/g) in the pre- and posttreatment as well as when the venom was preincubated with suramin. In addition, suramin also inhibited cultured endothelial cell lesion, as well as in vitro myotoxicity, evaluated in mouse extensor digitorum longus muscle, which was inhibited by suramin (10 and 25 µM), decreasing the rate of CK release, showing that suramin protected the sarcolemma against damage induced by components of bee venom (2.5 µg/mL). Moreover, suramin inhibited the in vivo myotoxicity induced by i.m. injection of A. mellifera venom in mice (0.5 µg/g). The analysis of the area under the plasma CK vs. time curve showed that preincubation, pre- and posttreatment with suramin (30 µg/g) inhibited bee venom myotoxic activity in mice by about 89%, 45% and 40%, respectively. Suramin markedly inhibited the PLA2 activity in a concentration-dependent way (1-30 µM). Being suramin a polyanion molecule, the effects observed may be due to the interaction of its charges with the polycation components present in A. mellifera bee venom.


Subject(s)
Antivenins/pharmacology , Bee Venoms/pharmacology , Muscle Fibers, Skeletal/drug effects , Suramin/pharmacology , Animals , Bee Venoms/antagonists & inhibitors , Capillary Permeability/drug effects , Cells, Cultured , Creatine Kinase/blood , Edema/chemically induced , Edema/drug therapy , Edema/pathology , Endothelium, Vascular/drug effects , Erythrocytes/drug effects , Evans Blue , Hematocrit , Injections, Intramuscular , Longevity/drug effects , Male , Mice , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/pathology , Phospholipases A2/metabolism , Rats , Sarcolemma/drug effects , Sarcolemma/enzymology , Skin/blood supply
15.
Purinergic Signal ; 9(1): 81-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22987361

ABSTRACT

Endothelial cells control vascular tone, permeability and leukocyte transmigration and are modulated by pro-inflammatory mediators. Schistosomiasis is an intravascular disease associated with inflammation, therefore altering endothelial cells' phenotype. Purinergic P2X7 receptors (P2X7R) play an important role in inflammation; however, the impact of the disease upon endothelial P2X7R function or expression has not been explored. Using ethidium bromide uptake to investigate P2X7R function, we observed that the effects of ATP (3 mM) and the P2X7R agonist 3'-O-(4-benzoyl)-ATP (BzATP) were smaller in mesenteric endothelial cells from the Schistosoma mansoni-infected group than in the control group. In the control group, BzATP induced endothelial nitric oxide production, which was blocked by the P2X7R antagonists KN-62 and A740003. However, in the infected group, we observed a reduced effect of BzATP and no effect of both P2X7R antagonists, suggesting a downregulation of endothelial P2X7R in schistosomiasis. We observed similar results in both infected and P2X7R(-/-) groups, which were also comparable to data obtained with KN-62- or A740004-treated control cells. Data from Western blot and immunocytochemistry assays confirmed the reduced expression of P2X7R in the infected group. In conclusion, our data show a downregulation of P2X7R in schistosomiasis infection, which likely limits the infection-related endothelial damage.


Subject(s)
Endothelium, Vascular/metabolism , Receptors, Purinergic P2X7/biosynthesis , Schistosomiasis mansoni/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Blotting, Western , Cell Membrane Permeability/drug effects , Cells, Cultured , Data Interpretation, Statistical , Fluorescent Dyes , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism
16.
PLoS One ; 6(8): e23547, 2011.
Article in English | MEDLINE | ID: mdl-21853150

ABSTRACT

BACKGROUND AND AIMS: Schistosomiasis is an intravascular parasitic disease associated with inflammation. Endothelial cells control leukocyte transmigration and vascular permeability being modulated by pro-inflammatory mediators. Recent data have shown that endothelial cells primed in vivo in the course of a disease keep the information in culture. Herein, we evaluated the impact of schistosomiasis on endothelial cell-regulated events in vivo and in vitro. METHODOLOGY AND PRINCIPAL FINDINGS: The experimental groups consisted of Schistosoma mansoni-infected and age-matched control mice. In vivo infection caused a marked influx of leukocytes and an increased protein leakage in the peritoneal cavity, characterizing an inflamed vascular and cellular profile. In vitro leukocyte-mesenteric endothelial cell adhesion was higher in cultured cells from infected mice as compared to controls, either in the basal condition or after treatment with the pro-inflammatory cytokine tumor necrosis factor (TNF). Nitric oxide (NO) donation reduced leukocyte adhesion to endothelial cells from control and infected groups; however, in the later group the effect was more pronounced, probably due to a reduced NO production. Inhibition of control endothelial NO synthase (eNOS) increased leukocyte adhesion to a level similar to the one observed in the infected group. Besides, the adhesion of control leukocytes to endothelial cells from infected animals is similar to the result of infected animals, confirming that schistosomiasis alters endothelial cells function. Furthermore, NO production as well as the expression of eNOS were reduced in cultured endothelial cells from infected animals. On the other hand, the expression of its repressor protein, namely caveolin-1, was similar in both control and infected groups. CONCLUSION/SIGNIFICANCE: Schistosomiasis increases vascular permeability and endothelial cell-leukocyte interaction in vivo and in vitro. These effects are partially explained by a reduced eNOS expression. In addition, our data show that the disease primes endothelial cells in vivo, which keep the acquired phenotype in culture.


Subject(s)
Cell Communication , Endothelial Cells/pathology , Leukocytes/pathology , Schistosomiasis/pathology , Animals , Caveolin 1/metabolism , Cell Adhesion , Cell Movement , Cells, Cultured , Endothelial Cells/enzymology , Male , Mesentery/pathology , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Peritoneal Cavity/pathology , Schistosoma mansoni/physiology , Schistosomiasis/enzymology
17.
Mem Inst Oswaldo Cruz ; 106(4): 456-60, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21739034

ABSTRACT

Schistosomiasis, classified by the World Health Organization as a neglected tropical disease, is an intravascular parasitic disease associated to a chronic inflammatory state. Evidence implicating inflammation in vascular dysfunction continues to mount, which, broadly defined, reflects a failure in the control of intracellular Ca2+ and consequently, vascular contraction. Therefore, we measured aorta contraction induced by 5-hydroxytryptamine (5-HT) and endothelin-1 (ET-1), two important regulators of vascular contraction. Isometric aortic contractions were determined in control and Schistosoma mansoni-infected mice. In the infected animals, 5-HT induced a 50% higher contraction in relation to controls and we also observed an increased contraction in response to Ca2+ mobilisation from sarcoplasmic reticulum. Nevertheless, Rho kinase inhibition reduced the contraction in response to 5-HT equally in both groups, discarding an increase of the contractile machinery sensitivity to Ca2+. Furthermore, no alteration was observed for contractions induced by ET-1 in both groups. Our data suggest that an immune-vascular interaction occurs in schistosomiasis, altering vascular contraction outside the mesenteric portal system. More importantly, it affects distinct intracellular signalling involved in aorta contraction, in this case increasing 5-HT receptor signalling.


Subject(s)
Aorta, Thoracic/drug effects , Endothelin-1/pharmacology , Receptors, Serotonin/drug effects , Schistosomiasis mansoni/physiopathology , Serotonin/pharmacology , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/parasitology , Aorta, Thoracic/physiopathology , Male , Mice , Schistosomiasis mansoni/immunology , Up-Regulation/drug effects
18.
Mem. Inst. Oswaldo Cruz ; 106(4): 456-460, June 2011. graf, tab
Article in English | LILACS | ID: lil-592189

ABSTRACT

Schistosomiasis, classified by the World Health Organization as a neglected tropical disease, is an intravascular parasitic disease associated to a chronic inflammatory state. Evidence implicating inflammation in vascular dysfunction continues to mount, which, broadly defined, reflects a failure in the control of intracellular Ca2+ and consequently, vascular contraction. Therefore, we measured aorta contraction induced by 5-hydroxytryptamine (5-HT) and endothelin-1 (ET-1), two important regulators of vascular contraction. Isometric aortic contractions were determined in control and Schistosoma mansoni-infected mice. In the infected animals, 5-HT induced a 50 percent higher contraction in relation to controls and we also observed an increased contraction in response to Ca2+ mobilisation from sarcoplasmic reticulum. Nevertheless, Rho kinase inhibition reduced the contraction in response to 5-HT equally in both groups, discarding an increase of the contractile machinery sensitivity to Ca2+. Furthermore, no alteration was observed for contractions induced by ET-1 in both groups. Our data suggest that an immune-vascular interaction occurs in schistosomiasis, altering vascular contraction outside the mesenteric portal system. More importantly, it affects distinct intracellular signalling involved in aorta contraction, in this case increasing 5-HT receptor signalling.


Subject(s)
Animals , Male , Mice , Aorta, Thoracic , Endothelin-1 , Receptors, Serotonin , Schistosomiasis mansoni , Serotonin , Vasoconstriction , Aorta, Thoracic , Aorta, Thoracic , Schistosomiasis mansoni/immunology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL